EP1594900A2 - Compositions et methodes de detection et de traitement des cancers a deficience de methylthioadenosine phosphorylase - Google Patents

Compositions et methodes de detection et de traitement des cancers a deficience de methylthioadenosine phosphorylase

Info

Publication number
EP1594900A2
EP1594900A2 EP04711185A EP04711185A EP1594900A2 EP 1594900 A2 EP1594900 A2 EP 1594900A2 EP 04711185 A EP04711185 A EP 04711185A EP 04711185 A EP04711185 A EP 04711185A EP 1594900 A2 EP1594900 A2 EP 1594900A2
Authority
EP
European Patent Office
Prior art keywords
mtap
antibody
protein
binding agent
sample
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP04711185A
Other languages
German (de)
English (en)
Inventor
Lorenzo M. Leoni
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Cephalon LLC
Original Assignee
Salmedix Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Salmedix Inc filed Critical Salmedix Inc
Publication of EP1594900A2 publication Critical patent/EP1594900A2/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/48Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving transferase
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57496Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving intracellular compounds
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/91Transferases (2.)
    • G01N2333/91091Glycosyltransferases (2.4)
    • G01N2333/91142Pentosyltransferases (2.4.2)

Definitions

  • the invention comprises binding agents against human methylthioadenosine phosphorylase (MTAP) protein, and uses thereof in determining the presence or absence of MTAP protein in an embedded sample, the prognosis of cancers associated with deletions of the gene encoding for MTAP protein, and methods of treatment for such cancers.
  • MTAP human methylthioadenosine phosphorylase
  • Methylthioadenosine phosphorylase is an enzyme found in all normal tissues that catalyzes the conversion of methylthioadenosine (MTA) into adenine and 5-methylthioribose-l -phosphate. Afterward, the adenine is salvaged to generate adenosine monophosphate, and the 5-methylthioribose-l -phosphate is converted to methionine and formate. Because of this salvage pathway, MTA can serve as an alternative purine source if de novo purine synthesis is blocked, e.g., with antimetabolites, such as L-alanosine.
  • MTAP deficiency is not only found in tissue culture cells but the deficiency is also present in primary leukemias, gliomas, melanomas, pancreatic cancers, non-small cell lung cancers (NSLC), bladder cancers, astrocytomas, osteosarcomas, head and neck cancers, myxoid chondrosarcomas, ovarian cancers, endometirial cancers, breast cancers, soft tissue sarcomas, non-Hodgkin lymphomas, and mesotheliomas (Kamatani et al. (1981) Proc. Natl. Acad.
  • the gene encoding for human MTAP maps to region 9p21 on human chromosome 9p. This region also contains the tumor suppressor genes pi 6 INK4 (also know as CDKN2A), an ⁇ pl5 INK4B These genes encode for pl6 and pi 5, which are inhibitors of the cyclin D-dependent kinases cdk4 and cdk6, respectively (Efferth et al. (2002) Blood, Cells, Molec, and Dis. 28:47-56; Kamp et al.(1994) Science 264:436-440; Harmon et al. (1994), Nature 371:257-261).
  • pl6 1NK4 ⁇ transcript can be alternatively spliced into a transcript encoding pl4 ARF .
  • pl4 ARF binds to MDM2 and prevents degradation of p53 (Pomerantz et al. (1998) Cell 92:713-723).
  • the 9p21 chromosomal region is of interest because it is frequently homozygously deleted in a variety of cancers, including leukemias, NSLC, pancreatic cancers, gliomas, melanomas, and mesothelioma.
  • the deletions often inactivate more than one gene.
  • Cairns et al. ((1995) Nat. Gen. 11 :210-212) reported that after studying more than 500 primary tumors, almost all the deletions identified in such tumors involved a 170 kb region containing MTAP,pl4 ARF and P16 INK4 ⁇ . Carson et al.
  • WO 99/67634 reported that a correlation exists between the stage of tumor development and loss of homozygosity of the gene encoding MTAP and the gene encoding pi 6. For example, deletion of the MTAP gene, but not pl6 INK4A was reported to be indicative of a cancer at an early stage of development, whereas deletion of the genes encoding for pi 6 and MTAP was reported to be indicative of a cancer at a more advanced stage of tumor development.
  • Garcia-Castellano et al. reported that in some osteosarcoma patients, the MTAP gene was present at diagnosis but was deleted at a later time point (Garcia-Castellano et al., supra).
  • Reference protein sequences for pl6 and alternative transcripts, including pl4, are deposited in GenBank under the following accession numbers NP_000068; NP_478102.1; NP_478103.1, and NP_478104.1.
  • Reference mRNA sequences for pi 6 and alternative transcripts, including pi 4 are. deposited in Genbank under accession numbers NM_000077.2; NM_058195.1; NM_058196.1; and NM_058197.1.
  • Reference protein sequences for pi 5 are deposited under GenBank accession numbers NP_004927.2 and NP_511042.1.
  • Reference mRNA sequences for pi 5 are deposited in GenBank under accession numbers NM_004936.2 and NM_078487.1.
  • the MTAP salvage pathway may offer an opportunity for selective tumor therapy which spares nomial tissues.
  • therapies based on treating MTAP deficient cancers with chemotherapeutic regimens that interfere with purine utilization is presently underway.
  • compositions and methods that identify MTAP deficient tumors (tumor cells that produce no or low amounts of MTAP protein) in biological samples, particularly those biological samples commonly used in a medical environment, such as formalin- fixed paraffin-embedded (FFPE) tissue specimens.
  • FFPE formalin- fixed paraffin-embedded
  • the ability to detect human MTAP protein using immunohistochemistry techniques may be advantageous over other immunoassay techniques, such as Western blotting, as well as oligonucleotide based procedures, such as Southern blotting, in that individual cells can be screened and the chance of contaminating tumor cells with normal cells is reduced.
  • MTAP-binding agents that specifically bind to human methylthioadenosine phosphorylase (MTAP) protein and methods of their use.
  • the invention is directed to MTAP-binding agents that bind to human MTAP in an embedded biological sample, including biological samples embedded in wax, nitrocellulose, polyethylene glycol, or plastic but not OCT compound. More preferred are MTAP-binding agents that bind to human MTAP protein in a biological sample embedded in paraffin. Even more preferred are MTAP- binding agents that bind to human MTAP in fixed biological samples embedded in paraffin. A preferred fixative is formalin.
  • Preferred MTAP-binding agents are antibodies that specifically bind to human MTAP, including polyclonal and monoclonal antibodies. Especially preferred are anti- MTAP monoclonal antibodies. Preferably, the antibodies bind to human MTAP protein with an affinity greater than c ⁇ 7 1
  • the invention is directed to the anti-MTAP monoclonal antibody produced by hybridoma cell line ATCC PTA-5001 and its use.
  • the invention is directed to monoclonal antibodies that bind to the same epitope as the monoclonal antibodies produced by hybridoma cell line ATCC PTA-5001.
  • the invention is directed to the hybridoma cell lines that produce anti-MTAP monoclonal antibodies.
  • the present invention is directed to a monoclonal antibody which is capable of binding human MTAP protein in an embedded biological sample wherein said biological sample is not embedded in OCT compound.
  • the monoclonal antibody binds to the same human MTAP epitope as the monoclonal antibody produced by a cell line having ATCC Accession No. PTA-5001.
  • the present invention is directed to a monoclonal antibody secreted by a cell line deposited by ATCC Accession No. PTA- 5001.
  • the present invention is directed to functional antigen binding fragments of a monoclonal antibody secreted by the cell line having ATCC Accession No. PTA-5001.
  • the present invention is directed to functional antigen binding fragments of a monoclonal antibody which binds to the same human MTAP epitope as a monoclonal antibody produced by a cell line having ATCC Accession No. PTA-5001.
  • the invention is directed to a method of making an isolated hybridoma that produces an antibody useful for assessing whether an embedded biological sample includes cells that contain human MTAP protein.
  • This method involves the following steps: (1) immunizing a mammal using a composition including a human MTAP polypeptide; (2) isolating splenocytes from the immunized mammal and fusing the isolated splenocytes with an immortalized cell to form hybridomas; and (3) screening the hybridomas for those that produce an antibody that specifically binds with human MTAP protein in an embedded biological sample.
  • the invention is directed to a method for detecting the presence or absence of human MTAP protein in an embedded biological sample, including samples embedded in wax, nitrocellulose, PEG, or plastic.
  • One disclosed method involves the following steps: (1) contacting the embedded biological sample with an MTAP-binding agent that forms a binding complex with human MTAP protein if present in the sample; and (2) detecting, using methods described herein, the quantity of binding complex formed. Detection of little to no binding complex is indicative of little or no human MTAP protein in the biological sample.
  • the sample is embedded in paraffin. Even more preferably, the sample is fixed.
  • the fixative is formalin.
  • Methods for detecting the binding complex include labeling the binding agent with a detectable marker.
  • the binding agent, contained in the binding complex can be detected by using a detectably labeled second agent that binds to the binding agent.
  • the binding agent, contained in the binding complex can be detected using an unlabeled second agent that binds to the binding agent. The second agent can then be detected using a detectably labeled third agent that binds to the second agent.
  • Another method, disclosed herein, for detecting the presence or absence of human MTAP protein in a formalin-fixed paraffin embedded biological sample includes the following steps: (1) heating the sample to melt the paraffin; (2) deparaffinizing the sample, (3) inducing epitope retrieval in the sample, (4) incubating the sample with an MTAP-binding agent that specifically binds with human MTAP protein to form a binding complex; and (5) detecting the binding complex formed. Detection of little to no binding complex is indicative of little or no human MTAP protein in the sample.
  • a preferred method for inducing epitope retrieval is tlirough the use of heat.
  • Another preferred method for epitope retrieval is heating followed by treatment with a proteolytic enzyme.
  • a preferred proteolytic enzyme is trypsin.
  • the invention is directed to methods of detecting the presence or absence of human MTAP protein in a sample including the following steps: (1) contacting the sample with a monoclonal antibody that specifically binds to human MTAP protein to form a binding complex; and (2) determining the presence or absence of binding complex in the sample, whereby the presence of the binding complex indicates the presence of human MTAP protein in the sample.
  • a preferred monoclonal antibody is the monoclonal antibody produced by hybridoma cell line ATCC PTA-5001.
  • the method is an immunoassay such as a Western blot or ELISA assay.
  • the invention is directed to an anti-human MTAP monoclonal antibody immobilized onto a solid surface.
  • the invention is directed to an MTAP-binding agent that specifically binds to human MTAP protein present in an embedded biological sample and yields a statistical score, based on staining intensities, that permits the identification of an embedded sample including cells homozygously deleted for the gene encoding human MTAP protein.
  • the embedded sample is embedded in wax, nitrocellulose, PEG, or plastic.
  • Preferred MTAP-binding agents for the method include antibodies. More preferred MTAP-binding agents for the method are monoclonal antibodies. Especially preferred are monoclonal antibodies produced by hybridoma cell line ATCC PTA-5001.
  • the methods disclosed herein can be used with embedded biological samples from patients with cancer.
  • the embedded sample is from a cancer that is selected from the group consisting of non-Hodgkin's lymphoma, mesothelioma, primary brain malignancies, (such as glioblastoma, glioma and astrocytoma), non-small cell lung cancer, leukemia, (such as acute lymphocytic leukemia, acute myelogenous leukemia, chronic lymphocytic leukemia, and chronic myelogenous leukemia), bladder cancer, pancreatic cancer, soft tissue sarcoma, osteosarcoma, or head and neck cancer.
  • the samples are fixed and embedded in paraffin.
  • Preferred MTAP-binding agents for use in the methods disclosed described herein are antibodies that specifically bind to human MTAP.
  • Preferred are polyclonal or monoclonal antibodies that specifically bind to human MTAP.
  • Preferable are antibodies that bind to human MTAP protein with an affinity greater than 10 5 M _1 and even more preferably with an affinity greater than 10 7 M "1 .
  • a preferred anti-human MTAP monoclonal antibody for use in the methods disclosed herein are the monoclonal antibodies produced by hybridoma cell line ATCC PTA-5001, or monoclonal antibodies that bind to the same epitope as the monoclonal antibodies produced by hybridoma cell ATCC PTA-5001.
  • the invention is directed to methods of selecting a patient for treatment of an MTAP deficient cancer with a therapy regimen directed to MTAP deficient cancers.
  • a preferred method includes the following steps: (1) providing an embedded biological sample of the cancer from said patient; (2) contacting said sample with an MTAP-binding agent that specifically forms a binding complex with human MTAP protein present in the sample; and (3) detecting the quantity of binding complex, as described herein. Using this method, cancers with low or no detectable binding complex indicate cancers amenable to treatments directed to MTAP-deficient cancers.
  • a preferred therapy regimen includes a drug that inhibits de novo purine synthesis.
  • Preferred de novo purine syntheis inhibitors include L-alanosine, 10-propargyl-lO-deazaaminopterin (PDX), N-(4-(((2,4-diamino- 6-pteridinyl)methyl)methylamino)benzoyl)-L-glutamic acid) (methofrexate), AG2037 (Agouron/Pfizer), 4-aminopteroylglutamic acid (aminopterin), 2,4-diamino-5-methyl- 6-[[(3,4,5-trimethoxyphenyl)amino]methyl]quinazoline (trimetrexate), pyritrexim, 10- ethyl-deaza-aminopterin (edatrexate), 4'-methylene-10-deazaaminopterin (MDAM), 10-propargyl-5,8-dideazafolic acid (PDDF), N-[5-[iV-(3,4-dihydro-2-methyl-4- ox
  • Suitable patients for screening according to these methods include patients with a non-Hodgkin's lymphoma, mesothelioma, primary brain malignancies, such as glioblastoma, glioma, and astrocytoma, non-small cell lung cancer, leukemia, such as ALL, AML, CLL and CML, bladder cancer, pancreatic cancer, soft tissue sarcoma, osteosarcoma, or head and neck cancer.
  • kits for determining whether an embedded biological sample contains human MTAP protein includes an MTAP-binding agent that specifically binds with human MTAP protein to form a binding complex, and an indicator capable of signaling the formation of said binding complex.
  • Preferred MTAP-binding agents included with the kits are antibodies, preferably polyclonal or monoclonal antibodies that specifically react with human MTAP. More preferred MTAP-binding agents are anti- MTAP monoclonal antibodies that are produced by hybridoma cell line ATCC PTA- 5001 or a monoclonal antibody that recognizes the same epitope as the monoclonal antibody produced by hybridoma cell line ATCC PTA-5001.
  • Also disclosed herein are methods for determining the homozygous deletion of the MTAP gene complex in a biological sample comprising the steps of: (1) contacting the sample with a binding agent that specifically binds to human MTAP protein to form a binding complex; and (2) detecting, as described herein, the quantity of binding complex in the sample. Detection of little to no binding complex is indicative of homozygous deletion of the MTAP gene complex.
  • the invention is directed to methods of treating a cancer patient with a therapeutically effective dose of a therapy regimen directed to an MTAP deficient cancer including the following steps: (1) providing an embedded biological sample from said patient; (2) contacting the embedded sample with a binding agent that specifically forms a binding complex with human MTAP protein in the embedded sample; (3) detecting the quantity of binding complex in the sample, whereby cancers with low or no detectable binding complex to human MTAP protein indicate MTAP deficient cancers amenable to treatment; and (4) administering a therapeutically effective amount of a therapy regimen directed to an MTAP deficient cancer.
  • a preferred therapy regimen includes a drug that inhibits de novo purine synthesis.
  • Preferred de novo purine synthesis inhibitors include L-alanosine, 10- propargyl-10-deazaaminopterin (PDX), N-(4-(((2 3 4-diamino-6- pteridinyl)methyl)methylamino)benzoyl)-L-glutamic acid) (methofrexate), AG2037 (Agouron Pfizer), 4-aminopteroylglutamic acid (aminopterin), 2,4-diamino-5-methyl- 6-[[(3,4,5-trimethoxyphenyl)amino]methyl]quinazoline (trimetrexate), pyritrexim, 10- ethyl-deaza-aminopterin (edatrexate), 4'-methylene-10-deazaaminopterin (MDAM), 10-propargyl-5,8-dideazafolic acid (PDDF), N-[5-[N-(3,4-dihydro-2-methyl-4- oxoquina
  • Preferred cancers for treatment include non-Hodgkin's lymphomas, mesotheliomas, glioblastomas, gliomas, non-small cell lung cancers, leukemias, bladder cancers, pancreatic cancers, soft tissue sarcomas, astrocytomas, osteosarcomas, head and neck cancers, or myxoid chondrosarcomas.
  • FIG. 1 depicts a Western blot of human MTAP protein using the antibodies produced by four subclones of hybridoma clone 6.
  • FIG. 2 depicts an immunohistochemistry blot of paraffin-embedded glioblastoma multiforme using anti-MTAP monoclonal antibody produced by hybridoma cell line PTA-5001.
  • MTAP-binding composition or "MTAP-binding agent” refer to molecules that bind with specificity to human MTAP protein or a fragment thereof.
  • the molecule may be a polymer, chemical reagent, an antibody, as defined herein, and other MTAP-binding proteins.
  • binding agent refers to a complex of an MTAP-binding agent and human MTAP protein.
  • Specific binding of the binding agent means that the binding agent has a specific binding site that recognizes a site on the human MTAP protein.
  • antibodies raised to the human MTAP protein and recognizing an epitope on the human MTAP protein are capable of forming a binding agentMTAP protein complex by specific binding.
  • the formation of a binding agentMTAP protein complex allows the detection of MTAP protein in a mixture of other proteins and biologies.
  • antibody MTAP protein complex refers to an MTAP-binding agentMTAP protein complex in which the MTAP-binding agent is an antibody.
  • the antibody may be monoclonal, polyclonal or even an antigen binding fragment of an antibody.
  • antigenic determinant refers to the specific portion of an antigen to which an antibody binds.
  • an "immunogenic epitope”, as used herein, is defined as a portion of a protein that elicits an antibody response in an animal, as dete ⁇ ined by any method known in the art, for example, by the methods for generating antibodies (See, for example, Geysen et al. (1983) Proc. Natl. Acad. Sci. USA 81 :3998-4002).
  • MTAP-binding agent such as an antibody, a protein, or peptide
  • an MTAP-binding agent such as an antibody, a protein, or peptide
  • an antibody is recognizing and binding to a specific protein structure rather than to proteins in general.
  • an antibody is specific for epitope "A”
  • the specificity can be demonstrated by a competition assay using labeled and unlabeled epitope A. Unlabeled epitope A will reduce the amount of labeled epitope A bound to the antibody.
  • Antibodies specific for a particular human MTAP epitope may recognize proteins highly similar to the MTAP protein.
  • the specified binding agents e.g., antibodies bind to a particular protein and do not significantly bind other proteins present in the sample. Specific binding to an antibody under such conditions may require an antibody that is selected for its specificity for a particular protein.
  • antibodies raised to the human MTAP protein immunogen with the amino acid sequence depicted in SEQ ID NO: 1 can be selected to obtain antibodies specifically immunoreactive with that MTAP protein and not with other proteins. These antibodies could recognize proteins highly similar to the human MTAP protein.
  • the term "antibody” refers to intact molecules as well as fragments thereof, such as Fab, F(ab') 2 , and Fv, which are capable of binding the antigenic determinant.
  • antibody in this specification is understood to include whole antibodies and useful fragments of antibodies.
  • Antibodies that bind human MTAP protein can be prepared using intact polypeptides or fragments containing small peptides of interest as the immunizing antigen.
  • the polypeptide or peptide used to immunize an animal can be derived from the translation of RNA or synthesized chemically, and can be conjugated to a carrier protein, if desired.
  • Commonly used carriers that are chemically coupled to peptides include bovine serum albumin and thyroglobulin.
  • the coupled peptide is then used to immunize the animal ⁇ e.g., a mouse, a rat, or a rabbit).
  • human MTAP protein when used in a protein context, means a protein having the amino acid sequence as shown in SEQ ID NO: 1 or a fragment of such a protein that can be used in an aspect of the present invention, e.g., used to raise antibody specific for human MTAP protein, used as a positive control to confirm binding of an MTAP-binding agent to human MTAP protein, and other uses.
  • use of the term "human MTAP protein” or “MTAP protein” in this specification is understood to include full-length MTAP protein, including trimeric and dimeric forms, and useful fragments of human MTAP protein.
  • a human MTAP protein may also be derivatized.
  • a human MTAP protein can interact with an MTAP-binding agent (as defined herein).
  • MTAP-binding agents e.g., antibodies
  • bind to the MTAP protein with high affinity e.g., at least about 100 nM, usually better than about 30 nM, preferably better than about 10 nM, and more preferably at better than about 3 nM.
  • polypeptide as used herein includes a fragment or segment of human MTAP protein, and includes a stretch of amino acid residues of at least about 8 amino acids, generally at least 10 amino acids, more generally at least 12 amino acids, often at least 14 amino acids, more often at least 16 amino acids, typically at least 18 amino acids, more typically at least 20 amino acids, usually at least 22 amino acids, more usually at least 24 amino acids, preferably at least 26 amino acids, more preferably at least 28 amino acids, and, in particularly preferred embodiments, at least about 30 or more amino acids.
  • a polypeptide of human MTAP can include all the amino acids represented in SEQ ID NO:l .
  • the term "monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to polyclonal antibody preparations that include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen.
  • biological sample is used herein in its broadest sense.
  • a biological sample suspected of containing human MTAP protein or a gene encoding for human MTAP protein may comprise one or more cells, an extract from cells, blood, tissue, and the like.
  • the term "fixed”, as used herein means treatment of a biological sample which results in preservation of histological detail. Such treatment can stabilize the proteins in the specimen, prevent changes to the sample caused by such things as mold, bacteria, and/or stops the continuation of enzyme metabolic processes (autolysis). Fixation can also change soluble substances within the cell to insoluble substances and protect the specimen from the denaturing effects of dehydrating agents and subsequent processing steps.
  • MTAP gene complex is the p 14-p 16-MTAP gene segment. This region comprises about a 200 kb region on chromosome 9p21-22.
  • an "immunoassay” is an assay that utilizes an antibody to specifically bind to the analyte ⁇ e.g., human MTAP protein).
  • the immunoassay is thus characterized by detection of specific binding of human MTAP protein or a fragment thereof to an antibody, i.e., detection of an antibody MTAP protein complex.
  • an immunoassay detects human MTAP protein using an antibody as an MTAP-binding agent, as opposed to the use of other physical or chemical properties to isolate, target, and quantify the analyte.
  • immunohistochemistry means a technique used to detect the presence of an antigen in histological samples.
  • effusion means an abnormal collection of fluid in a body cavity.
  • detecting the quantity of includes detection methods that result in an objective quantification of the entity to be detected ⁇ e.g., human MTAP protein or MTAP-binding complex), as well as subjective quantifications ⁇ e.g., intensity of staining).
  • objective quantifications include detecting the gram or milligram quantity of the entity.
  • subjective quantifications include detecting the presence or absence of an entity, or the relative presence or absence of an entity.
  • Such subjective quantifications may include the use of a scale, e.g., 0 to 4 pluses, with 0 indicating absence of the entity and 4+ indicating detection of a large quantity of the entity.
  • nucleic acid molecule and “polynucleotide” are used interchangeably in this application. These terms refer to any polyribonucleotide or polydeoxribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA. These terms are intended to include DNA molecules ⁇ e.g., cDNA) and RNA molecules ⁇ e.g., mRNA) and analogs of the DNA or RNA generated using nucleotide analogs.
  • a kit is any manufacture ⁇ e.g. a package or container) comprising at least one reagent, e.g., an MTAP-binding agent, for specifically detecting human MTAP.
  • the manufacture is preferably promoted, distributed, or sold as a unit for performing the methods of the present invention.
  • a therapeutically effective amount refers to that amount of the compound being administered which will relieve to some extent one or more of the symptoms of the disorder being treated.
  • a therapeutically effective amount refers to that amount which has the effect of (1) reducing the size of the tumor; (2) inhibiting (that is, slowing to some extent, preferably stopping) tumor metastasis; (3) inhibiting to some extent (that is, slowing to some extent, preferably stopping) tumor growth; and/or, (4) relieving to some extent (or, preferably, eliminating) one or more symptoms associated with the cancer.
  • the term "therapy regimen directed to an MTAP deficient cancer” means the use of a drug, combinations of drugs, or combinations of drugs and other cancer therapeutic measures, such as radiation therapy, which have been designed to take advantage of cancer cells having no or little MTAP protein.
  • DNA can be isolated from a genomic or cDNA library using labeled oligonucleotide probes having sequences identical or complementary to the sequences disclosed herein. Full-length probes may be used, or oligonucleotide probes may be generated by comparison of the sequences disclosed with other proteins and selecting specific primers. Such probes can be used directly in hybridization assays to isolate DNA encoding MTAP proteins, or probes can be designed for use in amplification techniques such as PCR, for the isolation of DNA encoding MTAP proteins.
  • cDNA is prepared from cells that express the MTAP protein.
  • cDNA is prepared from the mRNA and ligated into a recombinant vector.
  • the vector is transfected into a recombinant host for propagation, screening and cloning. Methods for making and screening cDNA libraries are well known. See Gubler et al. (1983) Gene 25:263-269; Sambrook et al., supra; or Coligan et al., supra.
  • the DNA can be extracted from tissue and either mechanically sheared or enzymatically digested to yield fragments of about 12-20 kb. The fragments are then separated by gradient centrifugation and cloned in bacteriophage lambda vectors. These vectors and phage are packaged in vitro, as described, e.g., in Sambrook et al., supra or Coligan et al., supra. Recombinant phage are analyzed by plaque hybridization as described in Benton et al. (1977) Science 196:180-182. Colony hybridization is carried out as generally described in, e.g. , Grunstein et al. (1975) Proc. Natl. Acad. Sci. USA 72:3961-3965.
  • DNA encoding an MTAP protein can be identified in either cDNA or genomic libraries by its ability to hybridize with the nucleic acid probes described herein, for example in colony or plaque hybridization experiments. The corresponding DNA regions are isolated by standard methods familiar to those of skill in the art. See Sambrook et al., supra.
  • Various methods of amplifying target sequences such as the polymerase chain reaction, can also be used to prepare DNA encoding MTAP proteins. Polymerase chain reaction (PCR) technology is used to amplify such nucleic acid sequences directly from mRNA, from cDNA, and from genomic libraries or cDNA libraries.
  • the isolated sequences encoding MTAP proteins may also be used as templates for PCR amplification.
  • oligonucleotide primers complementary to two 5' regions in the DNA region to be amplified are synthesized. The polymerase chain reaction is then carried out using the two primers. See Innis et al. (eds.) (1990) PCR Protocols: A Guide to Methods and Applications Academic Press, San Diego, Calif. Primers can be selected to amplify the entire regions encoding a selected full-length MTAP protein or to amplify smaller DNA segments as desired. Once such regions are PCR-amplified, they can be sequenced and oligonucleotide probes can be prepared from sequence obtained using standard techniques. The nucleotide sequence for the cDNA of human MTAP has been deposited with GenBank under accession number U22233 (SEQ ID NO:2).
  • Oligonucleotides for use as probes can be chemically synthesized according to the solid phase phosphoramidite triester method first described by Beaucage et al. (1983) Tetrahedron Lett. 22(20): 1859-1862, or using an automated synthesizer, as described in Needham-VanDevanter et al. (1984) Nucleic Acids Res. 12:6159-6168. Purification of oligonucleotides can be performed e.g., by native acrylamide gel electrophoresis or by anion-exchange HPLC as described in Pearson et al. (1983) J. Chrom. 255:137-149. The sequence of the synthetic oligonucleotide can be verified using the chemical degradation method of Maxam and Gilbert in Grossman and Moldave (eds.) (1980) Methods in Enzymology 65:499-560 Academic Press, New York.
  • DNAs which encode human MTAP protein or fragments thereof, can be obtained by chemical synthesis, screening cDNA libraries, or by screening genomic libraries prepared from a wide variety of cell lines or tissue samples.
  • These DNAs can be expressed in a wide variety of host cells for the synthesis of a full-length protein or fragments that can, e.g., be used to generate polyclonal or monoclonal antibodies; for binding studies; for construction and expression of modified molecules; and for structure/function studies.
  • Each of these human MTAP polypeptides can be expressed in host cells that are transformed or transfected with appropriate expression vectors. The polypeptides may be expressed as fusions with other proteins.
  • Expression vectors are typically self-replicating DNA or RNA constructs containing the desired MTAP gene or its fragments, usually operably linked to suitable genetic control elements that are recognized in a suitable host cell. These control elements are capable of effecting expression within a suitable host. The specific type of control elements necessary to effect expression will depend upon the eventual host cell used.
  • the genetic control elements can include a prokaryotic promoter system or a eukaryotic promoter expression control system, and typically include a transcriptional promoter, an optional operator to control the onset of transcription, transcription enhancers to elevate the level of mRNA expression, a sequence that encodes a suitable ribosome-binding site, and sequences that terminate transcription and translation.
  • Expression vectors also usually contain an origin of replication that allows the vector to replicate independently from the host cell.
  • the vectors described herein contain DNAs, which encode human MTAP protein or a fragment thereof, typically encoding, e.g., a biologically active polypeptide or protein.
  • the DNA can be under the control of a viral promoter and can encode a selection marker.
  • This invention includes the use of such expression vectors which are capable of expressing eukaryotic cDNA coding for human MTAP in a prokaryotic or eukaryotic host, where the vector is compatible with the host and where the eukaryotic cDNA coding for the protein is inserted into the vector such that growth of the host containing the vector expresses the cDNA in question.
  • expression vectors are designed for stable replication in their host cells or for amplification to greatly increase the total number of copies of the desirable gene per cell. It is not always necessary to require that an expression vector replicate in a host cell, e.g., it is possible to effect transient expression of the protein or its fragments in various hosts using vectors that do not contain a replication origin that is recognized by the host cell. It is also possible to use vectors that cause integration of an MTAP gene or its fragments into the host DNA by recombination, or to integrate a promoter that controls expression of an endogenous gene.
  • Vectors as used herein, comprise plasmids, viruses, bacteriophage, integratable DNA fragments, and other vehicles that enable the integration of DNA fragments into the genome of the host.
  • Expression vectors are specialized vectors that contain genetic control elements that effect expression of operably linked genes. Plasmids are the most commonly used form of vector but all other forms of vectors that serve an equivalent function are suitable for use herein. See, e.g., Pouwels et al. (1985 and Supplements) Cloning Vectors: A Laboratory Manual Elsevier, N.Y.; and Rodriquez et al. (eds.) (1988) Vectors: A Survey of Molecular Cloning Vectors and Their Uses Buttersworth, Boston, Mass.
  • Suitable host cells include prokaryotes, lower eukaryotes, and higher eukaryotes.
  • Prokaryotes include both gram-negative and gram-positive organisms, e.g., E. coli and B. subtilis.
  • Lower eukaryotes include yeasts, e.g., S. cerevisiae.
  • Higher eukaryotes include established tissue culture cell lines from animal cells, both of non-mammalian origin, e.g., insect cells, and birds, and of mammalian origin, e.g., human, primates, and rodents.
  • Prokaryotic host- vector s stems include a wide variety of vectors for many different species. As used herein, E. coli and its vectors will be used generically to include equivalent vectors used in other prokaryotes.
  • a representative vector for amplifying DNA is pBR322 or its derivatives.
  • Vectors that can be used to express human MTAP proteins or fragments include, but are not limited to, such vectors as those containing the lac promoter (pUC-series); tip promoter (pBR322-trp); Ipp promoter (the pIN-series); lambda-pp or pR promoters (pOTS); or hybrid promoters such as ptac (pDR540). See Brosius, et al.
  • Lower eukaryotes e.g., yeasts and Dictyostehum
  • yeast vectors typically consist of a replication origin (unless of the integrating type), a selection gene, a promoter, DNA encoding the desired protein or its fragments, and sequences for translation termination, polyadenylation, and transcription termination.
  • Suitable expression vectors for yeast include such constitutive promoters as 3-phosphoglycerate kinase and various other glycolytic enzyme gene promoters or such inducible promoters as the alcohol dehydrogenase 2 promoter or metallothionine promoter.
  • Suitable vectors include derivatives of the following types: self-replicating low copy number (such as the YRp-series), self-replicating high copy number (such as the YEp-series); integrating types (such as the Yip-series), or mini-chromosomes (such as the YCp- series).
  • Higher eukaryotic tissue culture cells may be used as host cells for expression of human MTAP protein.
  • most any higher eukaryotic tissue culture cell line may be used, e.g., insect baculovirus expression systems, whether from an invertebrate or vertebrate source.
  • mammalian cells are preferred to achieve proper processing, both cotranslationally and posttranslationally. Transformation or transfection and propagation of such cells are routine in the art.
  • Useful cell lines include HeLa cells, Chinese hamster ovary (CHO) cell lines, baby rat kidney (BRK) cell lines, insect cell lines, bird cell lines, and monkey (COS) cell lines.
  • Expression vectors for such cell lines usually include an origin of replication, a promoter, a translation initiation site, RNA splice sites (e.g. , if genomic DNA is used), a polyadenylation site, and a transcription termination site. These vectors also may contain a selection gene or amplification gene. Suitable expression vectors may be plasmids, viruses, or retroviruses carrying promoters derived, e.g., from such sources as from adenovirus, SV40, parvoviruses, vaccinia virus, or cytom ⁇ galovirus. Representative examples of suitable expression vectors include pCDNAl; pCD, see Okayama et al. (1985) Mol. Cell Biol. 5:1136-1142; pMClneo Poly-A, see Thomas et al. (1987) Cell 51:503-512; and a baculovirus vector such as pAC 373 or pAC 610.
  • the MTAP polypeptides or fragments thereof need not be glycosylated to elicit biological responses in certain assays.
  • the usual pattern will be that provided naturally by the expression system.
  • the pattern will be modifiable by exposing the polypeptide, e.g., in unglycosylated form, to appropriate glycosylating proteins introduced into a heterologous expression system.
  • a human MTAP gene may be co-transformed with one or more genes encoding mammalian or other glycosylating enzymes.
  • an azide process for example, an acid chloride process, an acid anhydride process, a mixed anhydride process, an active ester process (for example, p-nitrophenyl ester, N-hydroxysuccinimide ester, or cyanomethyl ester), a carbodiimidazole process, an oxidative-reductive process, or a dicyclohexylcarbodiimide (DCCD)/additive process can be used.
  • Solid phase and solution phase syntheses are both applicable to the foregoing processes.
  • the prepared protein and fragments thereof can be isolated and purified from the reaction mixture by means of peptide separation, for example, by extraction, precipitation, electrophoresis and various forms of chromatography, and the like. Purification can be accomplished by use of known protein purification techniques or by the use of the antibodies or binding partners herein described, e.g., in immunoabsorbant affinity chromatography. Immunoabsorbant affinity chromatography is carried out by first linking the antibodies to a solid support and contacting the linked antibodies with solubilized lysates of appropriate source cells, lysates of other cells expressing the protein, or lysates or supernatants of cells producing the proteins as a result of DNA techniques, see below.
  • Multiple cell lines may be screened for one which expresses the appropriate protein at a decreased or high level compared with other cells.
  • Human MTAP protein can be isolated from natural sources, or by expression from a transformed cell using an appropriate expression vector. Purification of the expressed protein is achieved by standard procedures, or may be combined with engineered means for effective purification at high efficiency from cell lysates or supernatants. FLAG or 6xHis tag segments can be used for such purification features.
  • the human MTAP protein amino acid sequence is provided in SEQ ID NO: 1.
  • the sequence allows preparation of peptides to generate antibodies to recognize such segments.
  • affinity reagents allow detection and purification of more protein, including full-length or recombinant forms.
  • An isolated MTAP gene DNA can be readily modified by nucleotide substitutions, nucleotide deletions, nucleotide insertions, and inversions of nucleotide stretches. These modifications result in novel DNA sequences which encode MTAP proteins, or fragments thereof having highly similar physiological, immunogenic, or antigenic activity as wild-type MTAP protein. Modified sequences can be used to produce mutant antigens or to enhance expression. Enhanced expression may involve gene amplification, increased transcription, increased translation, and other mechanisms. Such mutant MTAP protein derivatives include predetermined or site- specific mutations of the respective protein or its fragments.
  • “Mutant MTAP protein” encompasses a polypeptide having an amino acid sequence which differs from that of the human MTAP protein as found in nature, whether by way of deletion, substitution, or insertion. Generally, the variant will share many physicochemical and biological activities, e.g., antigenic or immunogenic, with wild-type human MTAP, and contain most or all of the disclosed sequence.
  • the present invention also includes recombinant proteins, e.g. , heterologous fusion proteins using segments from these proteins.
  • a heterologous fusion protein is a fusion of proteins or segments that are naturally not normally fused in the same manner.
  • the fusion product of an immunoglobulin with a respective MTAP polypeptide is a continuous protein molecule having sequences fused in a typical peptide linkage, typically made as a single translation product and exhibiting properties derived from each source peptide.
  • a similar concept applies to heterologous nucleic acid sequences.
  • Derivatives of these MTAP antigens include amino acid sequence mutants, glycosylation variants, and covalent or aggregate conjugates with other chemical moieties.
  • Covalent derivatives can be prepared by linkage of functionalities to groups which are found in the MTAP protein amino acid side chains or at the N- or C-termini, by means which are well known in the art. These derivatives can include, without limitation, aliphatic esters or amides of the carboxyl terminus, or of residues containing carboxyl side chains, O-acyl derivatives of hydroxyl group-containing residues, andN-acyl derivatives of the amino terminal amino acid or amino-group containing residues, e.g., lysine or arginine.
  • Acyl groups are selected from the group of alkyl-moieties including C3 to C18 normal alkyl, thereby forming alkanoyl aroyl species. Covalent attachment to carrier proteins may be important when immunogenic moieties are haptens.
  • glycosylation alterations are included, e.g., made by modifying the glycosylation patterns of a polypeptide during its synthesis and processing, or in further processing steps. Particularly preferred means for accomplishing this are by exposing the polypeptide to glycosylating enzymes derived from cells which normally provide such processing, e.g., mammalian glycosylation enzymes. Deglycosylation enzymes are also contemplated.
  • proteins comprising substitutions are encompassed, which should retain substantial immunogenicity, to produce antibodies which recognize a protein of SEQ ID NO: 1.
  • proteins which begin and end at structural domains will usually retain antigenicity and cross immunogenicity.
  • a major group of derivatives are covalent conjugates of the MTAP proteins or fragments thereof with other proteins or polypeptides. These derivatives can be synthesized in recombinant culture such as N- or C-terminal fusions or by the use of agents known in the art for their usefulness in cross-linking proteins through reactive side groups. Preferred protein derivatization sites with cross-linking agents are at free amino groups, carbohydrate moieties, and cysteine residues.
  • polypeptides may also have amino acid residues which have been chemically modified by phosphorylation, sulfonation, biotinylation, or the addition or removal of other moieties, particularly those which have molecular shapes similar to phosphate groups.
  • the modifications will be useful labeling reagents, or serve as purification targets, e.g., affinity ligands.
  • Other derivatives of human MTAP may involve covalent or aggregative association with chemical moieties. These derivatives generally fall into the three classes: (1) salts, (2) side chain and terminal residue covalent modifications, and (3) adsorption complexes, for example with cell membranes. Such covalent or aggregative derivatives are useful as immunogens, as reagents in immunoassays, or in purification methods such as for affinity purification of ligands or other binding ligands.
  • a human MTAP protein antigen can be immobilized by covalent bonding to a solid support such as cyanogen bromide-activated Sepharose, by methods which are well known in the art, or adsorbed onto polyolefin surfaces, with or without glutaraldehyde cross-linking, for use in an assay or purification of anti-MTAP protein antibodies.
  • the MTAP protein or fragements thereof can also be labeled with a detectable group, e.g., radioiodinated by the chloramine T procedure, covalently bound to rare earth chelates, or conjugated to another fluorescent moiety for use in diagnostic assays. Purification of human MTAP or fragments thereof may be effected by immobilized antibodies.
  • Various immunoassays may be used for screening to identify antibodies having the desired specificity. Numerous protocols for competitive binding or immunoradiometric assays using either polyclonal or monoclonal antibodies with established specificities are well known in the art. Such immunoassays typically involve the measurement of complex formation between human MTAP and its specific antibody. A two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfering human MTAP epitopes is preferred, but a competitive binding assay may also be employed (Maddox (1983) J. Exp. Med. 158:1211-1216).
  • Fragments which function as epitopes may be produced by any conventional means. (See, e.g., Houghten (1985) Proc. Natl. Acad. Sci. USA 82:5131- 5135, further described in U.S. Pat. No. 4,631,211).
  • antigenic epitopes preferably contain a sequence of at least 4 to 7, more preferably at least 8 to 40, and, most preferably, between about 15 to about 30 amino acids.
  • Preferred polypeptides comprising immunogenic or antigenic epitopes of human MTAP are at least 4, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 amino acid residues in length up to and including the complete amino acid sequence of the protein. Methods for obtaining these polypeptides are described herein. Many conventional techniques in protein biochemistry and immunology are used. Such techniques are well known and are explained in Mayer et al.
  • Non-limiting examples of antigenic polypeptides or peptides that can be used to generate MTAP-specific antibodies include: a polypeptide comprising, or alternatively consisting of, human MTAP amino acid residues (see, SEQ ID NO:l) from about amino acid residues 18-28, 75-85, 93-103, 143-150, 170-180, or 220-245.
  • “about” means the particularly recited ranges and ranges larger or smaller by several, a few, 10, 5, 4, 3, 2 or 1 amino acid residues at either or both the amino- and carboxy-termini.
  • These polypeptide fragments have been determined to bear antigenic epitopes of human MTAP polypeptide by the antigenicity analysis of Hopp et al. ((1981) Proc. Natl. Acad. Sci. USA 86:152-156).
  • Additional non-exclusive preferred antigenic epitopes include the antigenic epitopes disclosed herein, as well as portions thereof.
  • Preferred antigenic epitopes include the antigenic epitopes disclosed herein, as well as any combination of two, three, four, five or more of these antigenic epitopes.
  • Antigenic epitopes can be used as the target molecules in immunoassays. (See, for instance, Wilson et al. (1984) Cell 37:767-778; Sutcliffe et al. (1983) Science 219:660-666); Bittle et al. (1985) J. Gen. Virol. 66:2347-2354).
  • Epitope-bearing polypeptides of the present invention may be used to induce antibodies according to methods well known in the art including, but not limited to, in vivo immunization, in vitro immunization, and phage display methods. See, e.g., Sutcliffe et al., supra; Wilson et al., supra, and Bittle et al., supra.
  • Lfin vivo immunization animals may be immunized with free peptide; however, anti- peptide antibody titer may be boosted by coupling the peptide to a macromolecular carrier, such as keyhole limpet hemacyanin (KLH), albumin or tetanus toxoid.
  • KLH keyhole limpet hemacyanin
  • albumin tetanus toxoid.
  • peptides containing cysteine residues may be coupled to a carrier using a linker such as maleimidobenzoyl- N-hydroxysuccinimide ester (MBS), while other peptides may be coupled to carriers using a more general linking agent such as glutaraldehyde.
  • MFS maleimidobenzoyl- N-hydroxysuccinimide ester
  • the polypeptide is of sufficient length (at least about, 25 amino acids), the polypeptide may be presented without a carrier.
  • immunogenic epitopes comprising as few as 8 to 10 amino acids have been shown to be sufficient to raise antibodies capable of binding to, at the very least, linear epitopes in a denatured polypeptide ⁇ e.g., in Western blotting).
  • Animals such as rabbits, rats and mice are immunized with either free or carrier-coupled peptides, for instance, by intraperitoneal and/or intradermal injection of emulsions containing about 100 ⁇ g of peptide or carrier protein and Freund's adjuvant or any other adjuvant known for stimulating an immune response.
  • booster injections may be needed, for instance, at intervals of about two weeks, to provide a useful titer of anti-peptide antibody that can be detected, for example, by ELISA assay using free peptide adsorbed to a solid surface.
  • the titer of anti-peptide antibodies in serum from an immunized animal may be increased by selection of anti- peptide antibodies, for instance, by adsorption to the peptide on a solid support and elution of the selected antibodies according to methods well known in the art.
  • the human MTAP polypeptide or fragments thereof comprising an immunogenic or antigenic epitope can be fused to other polypeptide sequences.
  • MTAP polypeptides or fragments thereof may be fused with the constant domain of immunoglobulins (IgA, IgE, IgG, IgM), or portions thereof (CHI, CH2, CH3, or any combination thereof and portions thereof) resulting in chimeric polypeptides.
  • immunoglobulins IgA, IgE, IgG, IgM
  • CHI constant domain of immunoglobulins
  • CH2, CH3, or any combination thereof and portions thereof resulting in chimeric polypeptides.
  • Such fusion proteins may facilitate purification and may increase half- life in vivo. This has been shown for chimeric proteins consisting of the first two domains of the human CD4-polypeptide and various domains of the constant regions of the heavy or light chains of mammalian immunoglobulins.
  • IgG Fusion proteins that have a disulfide-linked dimeric structure due to the IgG portion disulfide bonds have also been found to be more efficient in binding and neutralizing other molecules than monomeric polypeptides or fragments thereof alone. See, e.g., Fountoulakis et al. (1995) J. Biochem., 270:3958-3964.
  • Nucleic acids encoding the above epitopes can also be recombined with a gene of interest as an epitope tag ⁇ e.g., the hemagglutinin ("HA") tag, 6xHis tag or flag tag) to aid in detection and purification of the expressed polypeptide.
  • an epitope tag e.g., the hemagglutinin ("HA") tag, 6xHis tag or flag tag
  • the gene of interest is subcloned into a vaccinia recombination plasmid such that the open reading frame of the gene is translationally fused to an amino-terminal tag consisting of six histidine residues.
  • the tag serves as a matrix binding domain for the fusion protein. Extracts from cells infected with the recombinant vaccinia virus are loaded onto Ni 2+ nitriloacetic acid-agarose column and histidine-tagged proteins can be selectively eluted with imidazole-containing buffers.
  • antibodies can be raised to human MTAP protein, including individual, polymorphic, allelic, strain, or species variants, and fragments thereof, both in their naturally occurring (full-length) forms and in their recombinant forms.
  • Anti-idiotypic antibodies may also be used.
  • Antibody fragments which contain specific binding sites for human MTAP, may also be generated.
  • fragments include, without limitation, the F(ab')2 fragments which can be produced by pepsin digestion of the antibody molecule and the Fab fragments which can be generated by reducing the disulfide bridges of the F(ab')2 fragments.
  • Fab expression libraries may be constructed to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity (Huse et al. (1989) Science 254:1275-1281).
  • Antibodies, including antigen binding fragments, specific for human MTAP or its fragments are useful in diagnostic applications to detect the presence or absence of human MTAP and/or its fragments.
  • Such diagnostic assays can employ lysates, live cells, fixed cells, cell cultures, cell extracts, body fluids, and further can involve the detection of antigens in serum, or the like.
  • Various commercial assays exist, such as radioimmunoassay (RIA), enzyme-linked immunosorbent assay (ELISA), enzyme immunoassay (EIA), enzyme-multiplied immunoassay technique (EMIT), substrate-labeled fluorescent immunoassay (SLFIA), and the like.
  • RIA radioimmunoassay
  • ELISA enzyme-linked immunosorbent assay
  • EIA enzyme immunoassay
  • EMIT enzyme-multiplied immunoassay technique
  • SFIA substrate-labeled fluorescent immunoassay
  • unlabeled antibodies can be employed by using a second antibody which is labeled and which recognizes the antibody to the human MTAP protein or to a particular fragment thereof.
  • the reagents may be useful for determining the presence or absence of MTAP in biological samples.
  • the assay may be directed to histological analysis of a biopsy, or evaluation of MTAP in a blood or tissue sample.
  • Recombinant protein is the preferred immunogen for the production of monoclonal or polyclonal antibodies.
  • Naturally occurring protein may also be used either in pure or impure form.
  • Synthetic peptides made using the human MTAP protein sequence described herein may also used as an immunogen for the production of antibodies to the human MTAP protein.
  • Recombinant protein can be expressed in eukaryotic or prokaryotic cells as described herein, and purified as described. The product is then injected into an animal capable of producing antibodies. Either monoclonal or polyclonal antibodies may be generated for subsequent use in immunoassays to measure the protein.
  • an immunogen preferably a purified protein
  • animals are immunized with the mixture.
  • the animal's immune response to the immunogen preparation is monitored by taking test bleeds and determining the titer of reactivity to the protein of interest, for example, using an ELISA assay.
  • ELISA assay When appropriately high titers of antibody to the immunogen are obtained, blood is collected from the animal and antisera are prepared. Further fractionation of the antisera to enrich for antibodies reactive to the protein can be done if desired. See, e.g., Harlow et al. (1988) Antibodies: A Laboratory Manual CSH Press.
  • Monoclonal antibodies may be obtained by various techniques familiar to those skilled in the art. Briefly, spleen cells from an animal immunized with a desired antigen are immortalized, commonly by fusion with a myeloma cell. See, e.g., Kohler and Milstein (1976) Eur. J. Immunol. 6:511-519, which is incorporated herein by reference. Alternative methods of immortalization include transformation with Epstein Barr Virus, oncogenes, or retroviruses, or other methods known in the art.
  • Colonies arising from single immortalized cells are screened for production of antibodies of the desired specificity and affinity for the antigen, and yield of the monoclonal antibodies produced by such cells may be enhanced by various techniques, including injection into the peritoneal cavity of a vertebrate host.
  • Antibodies including binding fragments and single chain versions, against predetermined fragments of human MTAP can be raised by immunization of animals with conjugates of the fragments with carrier proteins as described above. Monoclonal antibodies are prepared from cells secreting the desired antibody. These antibodies can be screened for binding to normal or mutant MTAP proteins, or screened for agonistic or antagonistic activity.
  • These monoclonal antibodies will usually bind with at least a K D of about less than 5x10 "4 M, 5x10 "5 M, 10 "5 M, 5x10 “6 M, 10 “6 M, 5x10 "7 M, 10 “7 M, 5x10 “8 M, 10 “8 M, 5x10 "9 M, 10 "9 M, 5x10 "10 M, 10 “10 M, 5xl0 “n M, 10 "11 M, 5xl0 "12 M, 10 "12 M, 5xl0 "13 M, 10 "13 M, 5xl0 "14 M, 10 '14 M.
  • Monoclonal antibodies may be prepared from various mammalian hosts, such as mice, rodents, primates, humans, etc. Description of techniques for preparing such monoclonal antibodies maybe found in, e.g., Stites, et al.
  • the population of hybridomas is then screened to isolate individual clones, each of which secretes a single antibody species to the immunogen.
  • the individual antibody species obtained are the products of immortalized and cloned single B cells from the immune animal generated in response to a specific site recognized on the immunogenic substance.
  • polypeptides and antibodies of the present invention may be used with or without modification, including chimeric or humanized antibodies. Frequently, the polypeptides and antibodies will be labeled by joining, either covalently or non-covalently, a substance that provides for a detectable signal.
  • labels and conjugation techniques are known and are reported extensively in both the scientific and patent literature. For instance, antibody molecules produced by a hybridoma can be labeled by metabolic incorporation of radioisotope-containing amino acids provided as a component in the culture medium.
  • Suitable labels include radionuclides, enzymes, substrates, cofactors, inhibitors, fluorescent moieties, chemiluminescent moieties, magnetic particles, and the like. Patents, teaching the use of such labels include U.S. Pat. Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241.
  • the antibodies of this invention can also be used for affinity chromatography in isolating human MTAP protein.
  • Columns can be prepared where the antibodies are linked to a solid support, e.g., particles, such as agarose, SEPHADEX, or the like, where a cell lysate may be passed through the column, the column washed, followed by increasing concentrations of a mild denaturant, whereby purified human MTAP protein will be released.
  • the antibodies may also be used to screen expression libraries for particular expression products. Usually the antibodies used in such a procedure will be labeled with a moiety allowing easy detection of presence of antigen by antibody binding.
  • Antibodies to human MTAP protein may be used for the analysis of, or identification of specific cell population components that express the respective protein. Further, antibodies to human MTAP may be used to screen biological samples, such as tumor specimens. Preferably, the biological samples are embedded and fixed, e.g., tumor biopsies embedded in paraffin and fixed with formalin. By assaying the expression products of cells that express or do not express human MTAP protein, it is possible to select patients that may benefit from treatments designed to take advantage of cancer cells that express low or no levels of MTAP protein.
  • deletions of MTAP are often associated with deletions of other genes on chromosome 9, such as pi 6 and pi 4, the determination that a tumor cell does not express MTAP can be used as a surrogate marker for pl6 and pl4 deletions. Deletions of pi 6 and pl4 in association with MTAP deletion are linked with advanced stage tumors.
  • the present invention includes antibodies that are capable of binding to the same antigenic determinant as the monoclonal antibody produced by hybridoma cell line ATCC PTA-5001, as described herein, or fragments thereof. Such antibodies would compete with the monoclonal antibodies produced by hybridoma cell line ATCC PTA-5001 for binding at that epitope.
  • class, isotype and other variants of the antibodies of the invention having the antigen-binding region of the antibody produced from hybridoma cell line PTA-5001 antibody can be constructed using recombinant class-switching and fusion techniques known in the art (see, e.g., Thammana et al. (1983) Eur. J Immunol. 13:614; Spira et al. (1984) J. Immunol. Meth. 74:307-15- Neuberger et al. (1984) Nature 312:604-608; and Oi et al. (1986) Biotechniques, 4(3):214-21).
  • chimeric antibodies or other recombinant antibodies ⁇ e.g., fusion proteins wherein the antibody is combined with a second protein such as a lymphokine or a tumor inhibitory growth factor) having the same binding specificity as the antibody produced from hybridoma cell line ATCC PTA- 5001 antibody fall within the scope of this invention.
  • a second protein such as a lymphokine or a tumor inhibitory growth factor
  • a particular protein can be measured by a variety of immunoassay methods.
  • immunoassay methods for a review of immunological and immunoassay procedures in general, see Stites et al. (eds.) (1991) Basic and Clinical Immunology (7th ed.).
  • the immunoassays of the present invention can be performed in any of several configurations, which are reviewed extensively in Maggio (ed.) (1980) Enzyme Immunoassay CRC Press, Boca Raton, Fla.; Tijan (1985) "Practice and Theory of Enzyme Immunoassays," Laboratory Techniques in Biochemisti ⁇ and Molecular Biology), Elsevier Science Publishers B.
  • immunoassay design considerations include preparation of antibodies (e.g., monoclonal or polyclonal) having sufficiently high-binding specificity for their antigen so the specifically bound antibody-antigen complex can be distinguished reliably from nonspecific interactions.
  • preferred binding specificity is such that the binding protein has a binding affinity for the marker protein of greater than about 10 5 M "1 preferably greater than about 10 7 M "1 .
  • Immunoassays for measurement of human MTAP protein can be performed by a variety of methods known to those skilled in the art. (see, e.g., U.S. Pat. Nos. 4,366,241; 4,376,110; 4,517,288; and 4,837,168). For a review of the general immunoassays, see also Asai (1993) Methods in Cell Biology Volume 37: Antibodies in Cell Biology, Academic Press, Inc. New York; Stites & Terr (1991) Basic and Clinical Immunology 7th Edition.
  • Immunological binding assays typically utilize a "capture agent" to specifically bind to and often immobilize the analyte (MTAP polypeptide).
  • the MTAP-binding agent is an antibody.
  • immunoassays to measure the protein can be competitive or noncompetitive.
  • Noncompetitive immunoassays are assays in which the amount of captured analyte (in this case, MTAP) is directly measured.
  • the binding agent ⁇ e.g., antibody
  • the binding agent is bound directly or indirectly to a solid substrate where it is immobilized.
  • immobilized MTAP antibodies capture MTAP protein present in a biological sample ⁇ e.g., a blood sample).
  • the MTAP protein thus immobilized is then bound by a labeling agent, such as a MTAP antibody bearing a label.
  • the second antibody may lack a label, but it may, in turn, be bound by a labeled third antibody specific to antibodies of the species from which the second antibody is derived. Free labeled antibody is washed away and the remaining bound labeled antibody is detected ⁇ e.g. , using a gamma detector where the label is radioactive).
  • the amount of analyte ⁇ e.g., MTAP) present in the sample is measured indirectly by measuring the amount of an added (exogenous) analyte displaced (or competed away) from a capture agent ⁇ e.g., MTAP antibody) by the analyte present in the sample.
  • a known amount of MTAP polypeptide is added to a sample with an unquantified amount of MTAP polypeptide, and the sample is contacted with a capture agent, e.g., an MTAP antibody that specifically binds MTAP.
  • the amount of added MTAP polypeptide that binds to the MTAP antibody is inversely proportional to the concentration of MTAP present in the test sample.
  • a homogeneous immunoassay may be performed in which a separation step is not needed.
  • the label on the protein is altered by the binding of the protein to its specific binding agent. This alteration in the labeled protein results in a decrease or increase in the signal emitted by label, so that measurement of the label at the end of the immunoassay allows for detection or quantitation of the protein.
  • Western blot analysis can be used to determine the presence or absence of human MTAP protein in a sample. Electrophoresis is carried out, e.g., on a tissue sample suspected of containing the protein. Following electrophoresis to separate the proteins, and transfer of the proteins to a suitable solid support such as a nitrocellulose filter, the solid support is incubated with an antibody reactive with the denatured protein. This antibody may be labeled, or alternatively may be it may be detected by subsequent incubation with a second labeled antibody that binds the primary antibody.
  • a preferred method, described herein, for determining the presence or absence of MTAP protein, in a biological sample, is immunohistochemistry (IHC).
  • Immunohistochemistry allows for the evaluation of micro-anatomical detail and heterogeneity in tissues and tumors. Immunohistochemistry is advantageous over other methods of analyses because it is the only method that can directly localize proteins to individual cell types. Differences among gene expression of normal and tumor tissue can be detected while simultaneously noting the changes in cell number and composition. In contrast, techniques, such as Western blotting require the use of cell extracts; therefore, a possibility exists of contamination of different cell types.
  • a primary MTAP-binding agent that recognizes human MTAP protein is introduced to a biological specimen.
  • the primary MTAP-binding agent can be, for example, selected from the group consisting of a chemical compound that specifically binds human MTAP, an antisera containing polyclonal antibodies specifically reactive with human MTAP protein, a monoclonal antibody that specifically binds with human MTAP protein, or antigen-binding fragments of monoclonal or polyclonal antibodies that specifically bind human MTAP protein.
  • a wash can be performed to remove unbound antibody.
  • a secondary antibody directed against the primary antibody and labeled with an enzyme, can be incubated with the biological specimen. During incubation, the secondary antibody will bind to the primary antibody.
  • the second antibody may lack a label, but it may, in turn, be bound by a labeled third antibody specific to antibodies of the species from which the second antibody is derived.
  • the primary MTAP-binding agent can be labeled with an enzyme thus eliminating the need for a second antibody.
  • the labeled MTAP-binding agent can be labeled with biotin rather than an enzyme. Then, in an additional step, enzyme-labeled avidin or streptavidin is introduced to the sample and allowed to bind to the biotinylated antibody.
  • the tissue sample may be fresh or frozen or may be embedded, for example, in paraffin or other waxes, nitrocellulose, carbowax (also known as water soluble polyethylene glycol ( see, Gao ed. (1993) "Polyethylene Glycol as an Embedment for Microscopy and Histochemistry," CRC Press, Inc. Boca Raton, FL), plastic, including resins such as acrylic and epoxy resins, or OCT embedded frozen blocks.
  • the samples are embedded in paraffin or other waxes, nictrocellulose, carbowax, or plastic.
  • the samples can be fixed with a preservative, such as formalin, for example.
  • a preferred embodiment of the invention is a formalin-fixed biological sample embedded in paraffin.
  • a preferred MTAP-binding agent for detecting the presence or absence of human MTAP protein in a biological sample is a monoclonal antibody the specifically binds to human MTAP protein.
  • Another preferred technique for determining the presence or absence of MTAP, using the antibodies described herein, is immunocytology. This technique is useful for the analysis of hematological cancers.
  • the MTAP-binding agents described herein are useful for determining the MTAP status of samples obtained from surgical biopsies, fine-needle biopsies, fine- needle aspiration biopsies, core-needle biopsies, effusions from body cavities, such as the abdominal cavity, the pleural cavities and the pericardial cavity, and cells collected from other bodily fluids, such as blood and urine and the like.
  • body cavities such as the abdominal cavity, the pleural cavities and the pericardial cavity
  • cells collected from other bodily fluids such as blood and urine and the like.
  • an effusion sample can be collected by puncturing the chest wall or abdominal wall with a needle and evacuating the fluid.
  • Samples from fine-needle aspirations, effusions or other bodily fluids can be spun onto slides using conventional centrifugation or a Cytospin® apparatus (Shandon, Runcorn, U.K.) or smeared onto an appropriate slide for staining and/or fixation.
  • Cell blocks can also be prepared from such samples by concentrating the cells contained therein. For example, cells can be concentrated, e.g., by centrifugation. After concentration, the cells can be fixed in a suitable fixing agent, such as formalin or alcohol and then embedded into paraffin or other suitable material as done for tissue in surgical pathology. Concentrated cells can also be processed for ThinPrep preparation using, for example, a Cytyc ThinPrep® processor (Cytyc Corp Boxborough, MA).
  • Fixation is an important step in immunohistochemical and immunocytochemical assays. Fixation stabilizes the proteins in the specimen and prevents changes caused by such things as mold, bacteria, and the continuation of enzyme metabolic processes (autolysis). Fixatives can also change soluble substances within the cell to insoluble substances. Fixatives also protect the specimen from the denaturing effects of dehydrating agents and subsequent processing steps.
  • Biological samples can be fixed using techniques known in the art. For example, air-drying can preserve blood smears. The smear can then be fixed using a fixative agent, such as, but not limited to, a high-grade methanol, anhydrous acetone, or formalin-based fixatives. Air-dried preparations may exhibit an overall lower MTAP antigen density, resulting in weak immunostaining. Thus, extended MTAP- binding agent incubation times may be needed.
  • a fixative agent such as, but not limited to, a high-grade methanol, anhydrous acetone, or formalin-based fixatives. Air-dried preparations may exhibit an overall lower MTAP antigen density, resulting in weak immunostaining. Thus, extended MTAP- binding agent incubation times may be needed.
  • Cytology smears may be fixed in 95% ethanol or spray-fixed with a carbowax containing alcoholic fluid.
  • Samples prepared using a Cytospin apparatus can be fixed using fixative agents such as acetone, alcohol, formalin, or paraformaldehyde.
  • Cryostat sections may be fixed using fixative agents such as alcohol or acetone.
  • biological samples may be fixed using, without limitation, formaldehyde-based fixatives, mercuric chloride-based fixatives, zinc- based fixatives, periodate-lysine-paraformaldehyde (PLP), glyoxal-based fixatives, Bouin's solution (contains formaldehyde, an aqueous saturated picric acid and glacial acetic acid), HoUande's Solution (contains cupric acetate that preserves red blood cell membranes, the granules of eosinophils and endocrine cells, and is capable of decalcifying small pieces of bone), ethanol, or acetone.
  • formaldehyde-based fixatives mercuric chloride-based fixatives
  • zinc- based fixatives zinc- based fixatives
  • periodate-lysine-paraformaldehyde (PLP) periodate-lysine-paraformaldehyde (PLP), glyoxal-based
  • decalcifying agents contain acid.
  • the acids used for decalcification may be either inorganic acids (hydrochloric and nitric) or organic acids (formic and acetic).
  • Chelating agents may also be used for decalcifying.
  • ethylenediaminetetraacetic acid (EDTA) is a preferred chelating agent for decalcification.
  • Alternate terminologies for "antigen retrieval” include “epitope retrieval,” “heat-induced epitope retrieval” (HIER), "target retrieval,” and “target unmasking.” Fixation may cause loss of immunoreactivity for many antigens.
  • the immunoreactivity of fixed tissue antigens and samples in cell blocks can be improved using techniques known in the art. For example, proteolytic digestion with proteolytic enzymes, such as, but not limited to, trypsin, bromelain, chymotrypsin, ficin, pepsin, or pronase, prior to adding the MTAP-binding agent can improve immunoreactivity.
  • suitable methods for epitope retrieval in embedded tissue sections and cell blocks include, heating, such as with, but not limited to, a microwave oven, autoclaves, steamers, water baths or pressure cookers.
  • Another suitable method for restoring immunoreactivity includes combining enzymatic digestion with heating in the presence of a retrieval solution such as, but not limited to, citrate buffer (about pH 6.0), EDTA buffer of about pH 8.0, or 0.01M TRIS-HC1 of pH about 1 or about 10.
  • a retrieval solution such as, but not limited to, citrate buffer (about pH 6.0), EDTA buffer of about pH 8.0, or 0.01M TRIS-HC1 of pH about 1 or about 10.
  • Antigen retrieval can be used for cytology slides, including Papanicolaou- stained slides. Heat induced antigen retrieval, if necessary, may be done using suitable methods, as described herein, such as water baths, pressure cookers, and microwave ovens and the like.
  • the immunoassay formats described above may employ labeled assay components, as described herein.
  • the label can be in a variety of forms.
  • the label may be coupled directly or indirectly to the desired component of the assay according to methods well known in the art.
  • a wide variety of labels may be used.
  • the component may be labeled by any one of several methods. Traditionally a radioactive label incorporating 3 H, 125 1, 35 S, 14 C, or 32 P is used.
  • Non-radioactive labels include ligands that bind to labeled antibodies, fluorophores, chemiluminescent agents, enzymes, and antibodies that can serve as specific binding pair members for a labeled protein.
  • Antibodies reactive with a particular protein can also be measured by a variety of immunoassay methods, as discussed herein.
  • immunoassay methods for reviews of immunological and immunoassay procedures applicable to the measurement of antibodies by immunoassay techniques, see, e.g., Stites and Terr (eds.) Basic and Clinical Immunology (7th ed.) supra; Maggio (ed.) Enzyme Immunoassay, supra; and Harlow and Lane Antibodies, A Laboratory Manual, supra.
  • FFPE formalin-fixed paraffin- embedded
  • the scoring system included an analysis of staining intensity.
  • the staining intensity of the test article was judged relative to the intensity of a control slide containing an adjacent section stained with a negative control antibody. Staining of the section labeled with the negative reagent control was considered “background.” A “0” indicates no staining relative to background. A “1+” indicates weak staining. A “2+” indicates moderate staining, and a “3+” indicates strong staining, (see, Table 1) Using standard pathology practice, staining intensity was reported at the highest level of intensity observed in all tissue elements, except the distinctive tissue element where an expanded scoring scheme was reported.
  • Short H-score (highest staining intensity observed x % of cells staining at that intensity).
  • a preferred long H score is about 50, more preferably about 20 and even more preferably about 5 or less.
  • MTAP-positive tissue controls the MTAP-expressing cell line A427 (ATCC), normal placental tissues and a tumor sample that was previously characterized as expressing MTAP were used. When possible, normal cells surrounding the tumor cells were also scored for MTAP positivity.
  • the data obtained using the anti-MTAP monoclonal antibody are in accord with previously reported data obtained in the same tumor types using hybridization based techniques to detect the presence or absence of the MTAP gene at the DNA or RNA levels.
  • mammalian hosts ⁇ e.g., humans) suffering from an MTAP deficient cancer may be treated with a therapeutically effective dose of drug, combinations of drugs, or drag and/or drugs and other cancer therapies (for example, radiation) determined to be of use in treating MTAP deficient cancers.
  • a therapeutically effective dose of drug, combinations of drugs, or drag and/or drugs and other cancer therapies for example, radiation
  • cancer therapies for example, radiation
  • One preferred class of drugs for treating MTAP deficient cancers are drugs that inhibit de novo purine synthesis, including IMPDH inhibitors.
  • Examples of such drugs are, without limitation, L-alanosine, 10- propargyl-10-deazaaminopterin (PDX), N-(4-(((2,4-diamino-6- pteridinyl)methyl)methylamino)benzoyl)-L-glutamic acid) (methofrexate), AG2037 (Agouron Pfizer), 4-aminopteroylglutamic acid (aminopterin), 2,4-diamino-5-methyl- 6-[[(3,4,5-trimethoxyphenyl)amino]methyl]quinazoline (trimetrexate), pyritrexim, 10- ethyl-deaza-aminopterin (edatrexate), 4'-methylene-10-deazaaminopterin (MDAM), 10-propargyl-5,8-dideazafolic acid (PDDF), N-[5-[N-(3,4-dihydro-2-methyl-4- oxoquinazolin-6
  • a preferred MTAP therapy regimen included in the scope of the invention is the use of "rescue" agents in combination with de novo purine synthesis inhibitors.
  • Rescue agents protect normal MTAP positive cells from exposure to cancer drags that are de novo purine synthesis inhibitors.
  • non-malignant, MTAP competent cells may be protected from any effect of exposure to de novo purine synthesis inhibitors through administration of MTA or a suitable substrate analogue for use in adenine synthesis.
  • Suitable compounds for use in this regard include MTA, 2'-5'-dideoxyadenosine, 5'-deoxyadenosine, 2'-deoxy-5-deoxy-5'methylthioadenosine.
  • MTAP competent cells are capable of producing adenine from metabolism of methylthioadenosine for replenishment of the AMP cellular pool and therefore would not be expected to be depleted of AMP to the same extent as MTAP deficient cells.
  • the present invention provides reagents which will find use in diagnostic applications as described elsewhere herein, or below in the description of kits for diagnosis.
  • Antibodies and other MTAP-binding agents directed towards human MTAP protein may be used to purify the corresponding MTAP protein.
  • Antibodies and other MTAP-binding agents may also be used in a diagnostic fashion to determine whether MTAP protein is present or absent in a tissue sample or cell population using the methods described herein.
  • many cancers are associated with a deletion of the gene encoding the MTAP protein, and therapies directed to MTAP deficient tumor cells are presently under development.
  • the MTAP-binding agents and methods described herein have use in identifying patients who would benefit from treatments designed to target MTAP deficient cancers.
  • MTAP deficient cells can be used as a surrogate marker for deletion of pl4 and pl6.
  • Tumors with deletions of the genes encoding pl4, pl6, and MTAP are associated with advanced stage tumors. Still further, because some cancers initially start off as MTAP positive but later become MTAP negative, that MTAP-binding agents described herein have use in monitoring tumor progression.
  • This invention also includes the use of the antibodies of the invention in a variety of diagnostic kits and methods for detecting the presence or absence of human MTAP protein in a biological sample.
  • a kit for determining the presence (high, normal or low expression) or absence of human MTAP protein in a sample would typically comprise an MTAP- binding agent, e.g., antibody, having known binding affinity for the human MTAP protein. Compartments containing reagents, and instructions, will normally be provided.
  • an MTAP- binding agent e.g., antibody
  • the reagents for diagnostic assays are supplied in kits, so as to optimize the sensitivity of the assay.
  • the protocol, and the label either labeled or unlabeled MTAP- binding agent may be included. This is usually in conjunction with other additives, such as buffers, stabilizers, materials necessary for signal production such as substrates for enzymes, and the like.
  • the kit will also contain instructions for proper use and disposal of the contents after use.
  • the kit has compartments for each useful reagent.
  • the reagents are provided as a dry lyophilized powder, where the reagents may be reconstituted in an aqueous medium providing appropriate concentrations of reagents for performing the assay.
  • labeling may be achieved by covalently or non-covalently joining a moiety that directly or indirectly provides a detectable signal.
  • the MTAP-binding agents e.g., antibodies can be labeled either directly or indirectly.
  • Possibilities for direct labeling include label groups such as radiolabels, e.g., I, enzymes (U.S. Pat. No. 3,645,090) such as peroxidase and alkaline phosphatase, and fluorescent labels (U.S. Pat. No. 3,940,475) capable of monitoring the change in fluorescence intensity, wavelength shift, or fluorescence polarization.
  • Possibilities for indirect labeling include biotinylation of one constituent followed by binding to avidin coupled to one of the above label groups.
  • the MTAP protein can be immobilized on various matrices followed by washing.
  • Suitable matrices include plastic such as an ELISA plate, filters, and beads.
  • Methods of immobilizing the MTAP protein to a matrix include, without limitation, direct adhesion to plastic, use of a capture antibody, chemical coupling, and biotin- avidin.
  • the last step in this approach involves the precipitation of protein/antibody complex by one of several methods including those utilizing, e.g., an organic solvent such as polyethylene glycol or a salt such as ammonium sulfate.
  • Other suitable separation techniques include, without limitation, the fluorescein antibody magnetizable particle method described in Rattle, et al. (1984) Clin. Chem. 30:1457- 1461, and the double antibody magnetic particle separation as described in U.S. Pat. No. 4,659,678.
  • Another diagnostic aspect of this invention involves use of the MTAP- binding agents of the invention in conjunction with oligonucleotide or polynucleotide probes based on the sequence of the polynucleotide encoding human MTAP protein.
  • the probes can be used for detecting polynucleotides that encode the .MTAP protein from samples of patients suspected of having an abnormal condition, e.g., cancer.
  • the preparation of both RNA and DNA nucleotide sequences, the labeling of the sequences, and the preferred size of the sequences has been discussed herein and in the literature.
  • an oligonucleotide probe should have at least about 14 nucleotides, usually at least about 18 nucleotides, and the polynucleotide probes may be up to several kilobases.
  • Preferred oligonucleotide probes to detect for the presence or absence of polynucleotides encoding MTAP protein include probes that detect exons 4, 5, or 8, or combinations of all three.
  • kits that also test for the qualitative or quantitative presence of other markers are also contemplated. Diagnosis or prognosis may depend on the combination of multiple indications used as markers. Thus, kits may test for combinations of markers, e.g., MTAP, pi 6, pl4 ARF . Such kits may contain a combination of reagents, for example, binding agents specific for MTAP, pi 6, or pl4 ⁇ RF , oligonucleotide probes specific for the polynucleotides that encode MTAP, pi 6, pl4 ⁇ RF , or combinations of all the aforementioned.
  • the cDNA sequence for MTAP is shown in SEQ ID NO:2.
  • the cDNA and protein sequences for pl6 and pl4 ARF are shown in SEQ ID NOS:3 through 10.
  • an immunohistochemical staining procedure can be employed that utilizes an MTAP- binding agent that selectively binds to human MTAP.
  • the sample to be screened can be embedded in a paraffin block or similar embedding material prior to the time the kit is used. Further, if the sample is not tissue but blood, plasma, or lymph, the sample can be fixed to a suitable surface, such as glass prior to the time the kit is used.
  • Hybridoma cell line 6.9 was deposited on February 11, 2003, with the American Type Culture Collection (ATCC), 10801 University Boulevard, Va. 20110- 2209 U.S.A and given ATCC patent deposit designation number PTA-5001.
  • ATCC American Type Culture Collection
  • Applicants' assignee, Salmedix, Inc. represents that the ATCC is a depository affording permanence of the deposit and ready accessibility thereto by the public if a patent is granted. All restrictions on the availability to the public of the material so deposited will be irrevocably removed upon the granting of a patent. The material will be available during the pendency of the patent application to one determined by the Commissioner to be entitled thereto under 37 CFR ⁇ 1.14, and 35 USC ⁇ 122.
  • the deposited material will be maintained with all the care necessary to keep it viable and uncontaminated for a period of at least five years after the most recent request for the furnishing of a sample of the deposited antibody, and in any case, for a period of at least thirty (30) years after the date of deposit or for the enforceable life of the patent, whichever period is longer.
  • Applicants' assignee acknowledges its duty to replace the deposit should the depository be unable to furnish a sample when requested due to the condition of the deposit.
  • Primers used for the amplification of MTAP were: sense 5'-CTC GCC CAC TGC AGA TTC CTT TCC CGT -3' (SEQ ID NO:l 1); antisense: 5'- GGC AGC CAT GCT ACT TTA ATG TCT TGG -3' (SEQ ID NO:12).
  • the PCR amplification was carried out in a 25 ⁇ l reaction mixture containing 1 ⁇ g of transcribed cDNA , lxPCR buffer [10 mM Tris-HCl (pH 8.0), 50 mM KC1, 1.5 mM MgCl 2 , and 0.0001% gelatin], 400 ⁇ M of each deoxynucleoside triphophate, 50 ng each of sense and antisense primers, and 2.5 units of Taq polymerase (Strategene, La Jolla, CA). Thirty cycles were performed with the programmable cyclic reactor (GeneAmp PCR system 9600; Perkin Elmer, Norwalk, CT). Each cycle consists of denaturation at 94°C for 1 min, annealing at 55°C for 1 min, and extension at 72°C for 1 min.
  • lxPCR buffer 10 mM Tris-HCl (pH 8.0), 50 mM KC1, 1.5 mM MgCl 2 , and 0.0001% gelatin
  • the MTAP cDNA was inserted into the expression vector pQE-32 (Qiagen).
  • the orientation of the insert was determined by digesting plasmid DNA with HindLLL.
  • the protein was purified as follows using the QIAexpress Expression System (Qiagen): E. coli strain M15[pREP4] was transformed with plasmid pQE-32 carrying human MTAP cDNA, and grown in 3L LB medium containing 100 ⁇ g/ml ampicillin and 25 ⁇ g/ml kanamycin with shaking until an OD 60 o of 0.434 was reached. Then, IPTG (isopropyl-beta-D-thiogalactopyranoside) was added to a final concentration of 2 mM and the culture was grown at 37°C with shaking for 5 hours. The cells were harvested by centrifugation at 4000 x g for 20 min.
  • QiAexpress Expression System Qiagen
  • the bacterial cell pellet was resuspended with 30 ml sonication buffer [50 mM NaH 2 PO 4 pH 8.0, 300 mM NaCl, 10 mM 2-mercaptoethanol, and 1 mM phenylmethylsulfonyl fluoride (PMSF)] and incubated on ice for 30 min. The suspension was frozen in liquid N 2 and stored overnight at -80°C.
  • 30 ml sonication buffer [50 mM NaH 2 PO 4 pH 8.0, 300 mM NaCl, 10 mM 2-mercaptoethanol, and 1 mM phenylmethylsulfonyl fluoride (PMSF)] and incubated on ice for 30 min.
  • the suspension was frozen in liquid N 2 and stored overnight at -80°C.
  • the column was washed with 100 ml sonication buffer at a flow rate less than 0.5 ml/min until the flow through OD 280 was less than 0.01 ml/min.
  • the column was washed with 160 ml wash buffer [50 mM NaH PO 4 pH 5.9, 1 M NaCl, 1 mM PMSF, 10%) glycerol].
  • Bound protein was eluted with 20 ml sonication buffer containing 0.5 M imidazole. The imidazole was removed and the protein concentrated using a Centriprep 10 (Amicon).
  • Fetal Bovine Serum 100 ml
  • NCTC 109 medium (Sigma N- 1140 or equivalent) 50 ml
  • ELISA REAGENTS 1. Carbonate-Bicarbonate Coating Buffer Capsule (Sigma).
  • pNPP p-Nitrophenol Phosphate
  • Dynex Immulon-4 plates 96-well plates, or 12-well removawell® strips and holder frame (Dynex, Chantilly, VA).
  • Screening ELISA plate preparation Purified recombinant MTAP protein (500 ng/well) was adsorbed onto microtiter plates in sodium carbonate/bicarbonate buffer overnight at 4°C, then blocked for nonspecific binding with phosphate- buffered saline (PBS) buffer containing 1%> bovine serum albumin (BSA). After blocking the plates were stored at -20 °C until use.
  • PBS phosphate- buffered saline
  • BSA bovine serum albumin
  • Diluted antiseram was added onto ELISA plates and incubated for 1 hr at room temperature. After incubation, the plates were washed 3 times with PBS to remove any unbound serum proteins. Alkaline-phosphatase conjugated anti-mouse secondary antibody was added to the plates, and incubation was continued at room temperature for 1 hr. At the end of the secondary antibody incubation, 3 washes with PBS were performed to remove non-specific secondary antibody. Then, the colorimetric substrate (p-nitrophenyl phosphate, PNPP) was added and the plates were analyzed after 5-30 min with a microtiter plate reader.
  • PNPP p-nitrophenyl phosphate
  • Recombinant MTAP protein (as described herein) was fractionated by 12%> sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and transferred onto a 0.22 ⁇ m-nitrocellulose membrane. After blotting, the membrane was blocked in Tris-buffered saline-Tween20 buffer (TBST) plus 5% non-fat dry milk (TBST-M) for 30 min at room temp. To determine the antibody specificity, antiseram was diluted in TBST-M and added to the nitrocellulose membrane. After 1 hr incubation at room temp, unbound antibody and serum proteins were removed by 3 washes with TBST. Alkaline-phosphatase (AP) conjugated anti-mouse secondary antibody was added to the membrane for additional 1 hour incubation and the protein signal was visualized by adding the AP substrate BCIP/NBT.
  • SDS-PAGE sodium dodecyl sulfate polyacrylamide gel electrophoresis
  • mice were immunized with the human MTAP protein produced above (including the His tag). Initially, each animal received 50 ⁇ g of purified recombinant protein mixed with complete Freund's adjuvant (CFA) by subcutaneous injection. Subsequent "boost" immunizations, using the same amount of antigen in incomplete Freund's adjuvant (IF A), were performed at 2-week intervals for a total of 4 boosts. After a total of 5 injections, the best responding mouse (as determined by analysis of polyclonal antiseram, as described below) was used for monoclonal antibody production.
  • CFA complete Freund's adjuvant
  • mice were given an additional immunization, without adjuvant, which was delivered by tail vein injection. Three days after the final injection, the mouse was sacrificed and the splenocytes were collected for hybridoma fusion as described herein.
  • Hybridoma cells were generated by the polyethylene glycol (PEG)-mediated cell fusion method. All solutions were pre-warmed to 37°C immediately prior to the fusion. Splenocytes from the immunized mouse and myeloma NS1 cells were collected by centrifugation and washed three times with DMEM. After the final wash, cells were combined in one tube and any trace amount of DMEM was removed by aspiration without disturbing the cell pellet. One milliliter of 50% PEG solution was added in a drop-wise fashion to the cell pellet over a one-minute interval with constant mixing.
  • PEG polyethylene glycol
  • the resulting hybridoma supernatants were screened primarily by ELISA, as described above, for reactivity with plates that had been previously coated with recombinant protein. Initially, 25 ELISA-positive clones were identified. These clones were propagated in DME-HT media and retested by ELISA to confirm the positive result and to eliminate any false-positive clones from the primary screen. After secondary screening, a total of 16 ELISA-positive hybridomas were identified. These positive hybridoma clones were expanded further in DME-HY media and the cells were preserved in liquid nitrogen for future single- cell cloning. The supernatants from these cultures were collected for further screening and characterization, as described below.
  • lysates from MTAP -expressing cells (the leukemia MOLT-4 [ATCC] and the lung cancer A427 [ATCC]) and MTAP- deleted cells (the leukemia JURKAT [ATCC] and the lung cancer A549 [ATCC]) were immunoblotted onto PVDF or nitrocellulose membranes.
  • the membranes were incubated with the supernatants from the hybridoma cultures (diluted 1:2 and 1 :10) and the reactive bands were revealed by chemiluminescence using secondary anti- mouse antibody conjugated to horseradish peroxidase (HRP) followed by incubation with chemiluminescent substrate.
  • HRP horseradish peroxidase
  • An MTAP-specific antibody would reveal an MTAP band at an approximate molecular weight of 30-35 kDa only in the MOLT-4 and A427 lanes, but not in the lanes containing JURKAT or A549 lysates.
  • Using whole cell lysates to identify MTAP-specific antibodies has the advantage of allowing the detection of possible cross-reactivity with unrelated proteins. The best supernatant identified using this screening was clone 6.
  • Clone 6 was selected for single-cell cloning to generate anti-MTAP monoclonal antibodies.
  • Parental hybridoma cells from clone 6 were grown from frozen stock and the supernatant was collected and tested again by ELISA. The serial-dilution method was used for single-cell cloning. Briefly, hybridoma cells were counted and plated out onto a total often 96-well plates at a concentration of 1, 0.5, and 0.1 cells per well.
  • Clones grown from a single cell were initially screened again by ELISA against purified MTAP protein, and then tested by immunoblotting in MTAP-expressing and MTAP-deleted cells, as described above to ensure the antibody's specificity and reactivity as seen with the parental hybridoma.
  • a total of 4 sub-clones derived from the parental clone 6 were chosen after screening by Western blot analysis (clones 6.9, 6.11, 6.22 and 6.23) (Fig.l). Each of these four sub-clones was expanded in culture to a larger quantity and then frozen in liquid nitrogen to preserve the antibody producing cell line. The tissue culture supernatant was collected for final testing using the described immunoblotting technique. Clone 6.9 was identified as having acceptable properties.
  • Tissue culture supernatant was collected from clone 6.9 and the anti-MTAP monoclonal antibody was purified by protein G-affinity chromatography. Briefly, the supernatant was cleared of cellular debris by high-speed centrifugation followed by ultra-filtration (0.22 ⁇ m filter) before being loaded onto a protein-G column. After antibody binding, the column was washed with PBS to remove any non-specific antibody and unbound proteins that were present in the supernatant. The anti-MTAP monoclonal antibody was eluted by 0.1 M glycine solution (pH 2.8) and the antibody- containing fractions were pooled and dialyzed extensively against PBS at 4°C. The final purified antibody concentration was determined by UV (280nm) absorbance.
  • Example 4 Immunohistochemistry
  • Mouse monoclonal anti-MTAP antibody, clone 6.9, of human LgGi isotype was stored at -85°C at a stock concentration of 3.3 mg/ml. Aliquots of the monoclonal antibody were stored at 2-8°C for up to four weeks.
  • Negative control for the mouse monoclonal anti-MTAP antibody was mouse IgGik (Sigma-Aldrich). The antibody was received on dry ice at a stock concentration of lmg/ml, and aliquots were stored at -20°C. Aliquots of 40 ⁇ l were stored at -20°C. When thawed, they were stored at 2-8°C for up to one week.
  • MTAP is present in all normal tissues, the ideal control tissue would be one in which the gene had been deleted.
  • two non-small cell carcinomas were selected: one in which MTAP was present in the cells and detected via immunohistochemistry; and one in which MTAP was deleted from the cells and not detected via immunohistochemistry.
  • An additional positive control included MTAP-expressing human non-small cell lung carcinoma A427 (ATCC) embedded in a paraffin block.
  • the negative reagent control is a species- and isotype-matched antibody ran at the same concentration on the same tissues as the primary antibody.
  • a pretreatment analysis was performed using non-small cell lung carcinoma as the tissue control and the positive and negative control cell lines. In this analysis, the following pretreatments were tested using 10 ⁇ g/ml of the mouse monoclonal MTAP antibody:
  • H.I.E.R. Pressure Cooker, BORG
  • trypsin for one minute was selected as a suitable pretreatment and used for the remainder of the study.
  • the MTAP binding-agent titer is selected as the lowest concentration that produces the highest combination of staining intensity and percentage of positively staining cells while minimizing background staining. Since the purpose of the assay is to detect the absence of a protein as the result of a gene deletion, a higher titer of MTAP-binding agent was selected to minimize possible false negatives. A suitable titer for this test was selected as the highest concentration of antigen binding agent that produces strongest staining while minimizing isotype and stromal background staining.
  • a titration analysis was performed with the mouse anti-MTAP monoclonal antibody on fixed, paraffin-embedded specimens. Initially, five-serial dilutions (20 ⁇ g/ml, 10 ⁇ g/ml, 5 ⁇ g/ml, 2.5 ⁇ g/ml, 1.25 ⁇ g/ml) were tested on the positive and negative cell line controls as well as the tissue control (placenta). Using a non-small cell carcinoma cell line deleted for MTAP, a titration analysis was performed using five-serial dilutions (80 ⁇ g/ml, 40 ⁇ g/ml, 20 ⁇ g/ml, 10 ⁇ g/ml, and 5 ⁇ g/ml). Based on these studies, a primary antibody (and negative reagent control) titer of 20 ⁇ g/ml was selected for the remainder of the study.
  • mice monoclonal MTAP antibody All immunohistochemical testing for the mouse monoclonal MTAP antibody was performed using the Mouse EnVision+TM Kit (DAKO Corporation, Carpinteria, CA) according to manufacturer's instructions.
  • DAKO Corporation Carpinteria, CA
  • paraffin-embedded specimens slides containing microtome-cut sections were removed from incubation in a 60°C oven after at least 60 minutes and cooled to room temperature. The slides were deparaffinized in xylene and graded alcohols, rinsed in running water, and rinsed three times in deionized water.
  • the slides were then incubated in a pressure cooker (BORG) at 120°C for 3 minutes followed by trypsin for one minute, and were then washed three times in PBS (Phosphate-Buffered Saline; DAKO Corp.). Endogenous peroxidase activity was blocked with a 5-minute incubation in a hydrogen peroxide solution contained in the EnVision+TM Kit, followed by tliree PBS washes. The slides were then incubated with the primary antibody (or the appropriate negative reagent control) at 20 ⁇ l/ml for 30 minutes at room temperature.
  • BORG pressure cooker
  • PBS Phosphate-Buffered Saline
  • a competitive enzyme immunoassay is carried out to detect the epitope overlap of an antibody with the monoclonal antibody produced by hybridoma cell line ATCC PTA-5001.
  • MTAP protein as described herein, is biotinylated with D-biotinyl- ⁇ -amidocaproic acid-N-hydroxysuccinimide ester (Roche Diagnostics GmbH, Catalogue No. 1 008 960) according to the manufacturer's instructions. 300 ng of this biotinylated antigen is bound in a volume of 100 ⁇ l PBS to a streptavidin-coated microtiter plate (produced according to EP-A 0 344 578) by incubating for 1 hour at room temperature.
  • Recombinant human MTAP protein is digested with various proteolytic enzymes (in individual experiments). For trypsin digestion, partial digestion of recombinant human MTAP protein is done in 50 mM Tris-HCl, 2 mM CaCl 2 , pH 8.0 at 37°C for 4 hr with an enzyme to substrate ratio of 1 : 1. The time of digestion is determined by monitoring the amount of digestion at various time points. The digestion products and controls are ran on both non-reducing and reducing (as described above) SDS-PAGE. For non-reducing gels, fragments obtained after proteolytic digestion are ran on 12.5%o (w/v) native polyacrylamide gels. After separation, the recombinant protein and digested fragments are transferred onto a 0.22 ⁇ m-nitrocellulose membrane. Blots are probed with anti-MTAP monoclonal antibody 6.9 as described above.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Cell Biology (AREA)
  • Biochemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Analytical Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Food Science & Technology (AREA)
  • General Physics & Mathematics (AREA)
  • Hospice & Palliative Care (AREA)
  • Genetics & Genomics (AREA)
  • Oncology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Toxicology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

L'invention concerne des compositions et des méthodes faisant appel à des agents se liant spécifiquement à une protéine de méthylthioadénosine phosphorylase (MTAP). Les compositions de cette invention contiennent des agents liants se liant à une protéine humaine MTAP dans des échantillons biologiques, y compris des échantillons inclus. Les agents liants sont utiles, par exemple, dans la détection, le pronostic et/ou le traitement de cancer à déficience de MTAP. Font également l'objet de cette invention des kits contenant les réactifs nécessaires à la détection d'une protéine humaine MTAP dans un échantillon inclus.
EP04711185A 2003-02-14 2004-02-13 Compositions et methodes de detection et de traitement des cancers a deficience de methylthioadenosine phosphorylase Withdrawn EP1594900A2 (fr)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US44788803P 2003-02-14 2003-02-14
US447888P 2003-02-14
US46071503P 2003-04-04 2003-04-04
US460715P 2003-04-04
PCT/US2004/004371 WO2004074325A2 (fr) 2003-02-14 2004-02-13 Compositions et methodes de detection et de traitement des cancers a deficience de methylthioadenosine phosphorylase

Publications (1)

Publication Number Publication Date
EP1594900A2 true EP1594900A2 (fr) 2005-11-16

Family

ID=32912275

Family Applications (1)

Application Number Title Priority Date Filing Date
EP04711185A Withdrawn EP1594900A2 (fr) 2003-02-14 2004-02-13 Compositions et methodes de detection et de traitement des cancers a deficience de methylthioadenosine phosphorylase

Country Status (5)

Country Link
US (1) US7157551B2 (fr)
EP (1) EP1594900A2 (fr)
CA (1) CA2516191A1 (fr)
TW (1) TW200427463A (fr)
WO (1) WO2004074325A2 (fr)

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1814544A4 (fr) * 2004-11-05 2009-12-02 Cephalon Inc Traitements anti-cancereux
US8436190B2 (en) 2005-01-14 2013-05-07 Cephalon, Inc. Bendamustine pharmaceutical compositions
WO2006113536A2 (fr) * 2005-04-14 2006-10-26 Hana Biosciences, Inc. Utilisation de pt523 pour le traitement des cancers
WO2009014642A1 (fr) * 2007-07-19 2009-01-29 Amgen Inc. Combinaisons pour le traitement du cancer
WO2009032057A2 (fr) 2007-08-29 2009-03-12 Adam Lubin Procédé pour la thérapie sélective d'une maladie
AR072777A1 (es) 2008-03-26 2010-09-22 Cephalon Inc Formas solidas de clorhidrato de bendamustina
CA2735899A1 (fr) 2008-09-25 2010-04-01 Cephalon, Inc. Formulations liquides de bendamustine
WO2010083276A1 (fr) * 2009-01-15 2010-07-22 Cephalon, Inc. Nouvelles formes de base libre de bendamustine
JP2020510618A (ja) * 2016-12-01 2020-04-09 グラクソスミスクライン、インテレクチュアル、プロパティー、ディベロップメント、リミテッドGlaxosmithkline Intellectual Property Development Limited 癌を処置する方法
JP7016958B2 (ja) * 2017-12-21 2022-02-07 ボード オブ リージェンツ,ザ ユニバーシティ オブ テキサス システム アデノシンおよび/またはメチルチオアデノシンの酵素媒介枯渇方法
CN115244175A (zh) 2020-01-07 2022-10-25 得克萨斯大学体系董事会 用于癌症治疗的改进的人甲硫腺苷/腺苷消耗酶变体
CN114515294A (zh) 2022-02-25 2022-05-20 浙江中医药大学 5’-甲基硫代腺苷在制备肥胖抑制药物或保健品中的应用

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4376110A (en) 1980-08-04 1983-03-08 Hybritech, Incorporated Immunometric assays using monoclonal antibodies
US4366241A (en) 1980-08-07 1982-12-28 Syva Company Concentrating zone method in heterogeneous immunoassays
US4517288A (en) 1981-01-23 1985-05-14 American Hospital Supply Corp. Solid phase system for ligand assay
US4569788A (en) * 1983-05-18 1986-02-11 The United States Of America As Represented By The Department Of Health And Human Services Monoclonal antibodies against non small cell lung cancer
US4877168A (en) * 1985-09-20 1989-10-31 Bott John Anthony Vehicle article carrier
CA1291031C (fr) 1985-12-23 1991-10-22 Nikolaas C.J. De Jaeger Methode pour la detection de liants specifiques et des substances liables par ceux-ci
US5840505A (en) 1993-12-29 1998-11-24 The Regents Of The University Of California Method for inhibiting adenylosuccinate synthetase activity in methylthioadenosine phosphorylase deficient cells
US5571510A (en) 1993-12-29 1996-11-05 The Regents Of The University Of California Method for selective methionine starvation of malignant cells in mammals
CZ289155B6 (cs) 1993-12-29 2001-11-14 The Regents Of The University Of California Izolovaný polynukleotid, který kóduje methylthioadenosin fosforylázu, expresní vektor a protilátka
US6214571B1 (en) 1993-12-29 2001-04-10 The Regents Of The University Of California Method for inhibiting adenylosuccinate synthetase activity in malignant methylthioadenosine phosphorylase deficient cells
US5942393A (en) 1993-12-29 1999-08-24 The Regents Of The University Of California Method for the detection of the presence or absence of methylthioadenosine phosphorylase (MTASE) in a cell sample by detection of the presence or absence of MTASE encoding nucleic acid in the cell sample
US6210917B1 (en) 1993-12-29 2001-04-03 The Regents Of The University Of California Method for suppressing multiple drug resistance in cancer cells
CH689642A5 (fr) * 1994-02-18 1999-07-30 Speno International Installation pour le reprofilage des rails d'une voie ferrée.
US5744585A (en) * 1995-03-16 1998-04-28 Medenica; Rajko D. Human monoclonal antibody against lung carcinoma
US6576420B1 (en) 1998-06-23 2003-06-10 Regents Of The University Of California Method for early diagnosis of, and determination of prognosis in, cancer
WO2004012769A1 (fr) * 2002-08-02 2004-02-12 The Regents Of The University Of California Inhibition therapeutique des proteines kinases dans des cellules cancereuses

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2004074325A2 *

Also Published As

Publication number Publication date
WO2004074325A2 (fr) 2004-09-02
US7157551B2 (en) 2007-01-02
WO2004074325A3 (fr) 2005-01-27
TW200427463A (en) 2004-12-16
CA2516191A1 (fr) 2004-09-02
US20040247600A1 (en) 2004-12-09

Similar Documents

Publication Publication Date Title
AU2008207533B2 (en) Methods for diagnosis and treatment of epithelial-derived cancers
US20070207510A1 (en) Methods and compounds for lymphoma cell detection and isolation
US9920135B2 (en) Anti-human CD26 monoclonal antibody or antigen-binding fragment thereof
KR101393144B1 (ko) 요로상피암의 검출용 키트 및 방법
KR20110052665A (ko) 암의 검출 방법
US20090123461A1 (en) Human podocalyxin alternative-spliced forms and uses thereof
US7157551B2 (en) Compositions and methods for the detection and treatment of methylthioadenosine phosphorylase deficient cancers
US7531314B2 (en) Therapeutic and diagnostic applications of perlecan domain I splice variants
US6432636B1 (en) Diagnostic applications of perlecan domain I splice variants
WO1998008381A9 (fr) Applications therapeutiques et diagnostiques d'alleles produits par epissure dans le domaine i du perlecan
EP0642664A1 (fr) Procedes de recherche de o?6 -methylguanine-adn methyltransferase
JP4532273B2 (ja) 癌に関係するタンパク質
CA2422538C (fr) Agonistes du recepteur notch
JP5339676B2 (ja) 細胞を検出する方法、およびこれに有用な薬剤
WO2009039584A1 (fr) Diagnostic et traitement d'un tissu atteint de maladie et lésé
JP5354634B2 (ja) ヒトabh8タンパク質、それをコードする遺伝子、およびこれらの治療的又は診断的用途
US7411049B2 (en) Hybridoma cell lines and monoclonal antibodies recognizing Prox1
KR101567053B1 (ko) 방사선 피폭에 의한 간 손상 예측용 바이오마커 및 그 예측방법
JP6537095B2 (ja) 癌の再発及び/又は転移の予測診断用マーカー
JP3230220B2 (ja) ブタ由来インターロイキン−18に対するモノクローナル抗体
JP4829961B2 (ja) プロスタシン部分ペプチド及び抗プロスタシン抗体
JPS62500584A (ja) モノクロ−ナル抗体およびその用途
JP2002308900A (ja) 抗ヒト肝性トリグリセリドリパーゼ抗体

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20050914

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LI LU MC NL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL LT LV MK

DAX Request for extension of the european patent (deleted)
RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: CEPHALON, INC.

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN

18W Application withdrawn

Effective date: 20061228